TY - JOUR A1 - Derakhshani, Shaghayegh A1 - Kurz, Andreas A1 - Japtok, Lukasz A1 - Schumacher, Fabian A1 - Pilgram, Lisa A1 - Steinke, Maria A1 - Kleuser, Burkhard A1 - Sauer, Markus A1 - Schneider-Schaulies, Sibylle A1 - Avota, Elita T1 - Measles Virus Infection Fosters Dendritic Cell Motility in a 3D Environment to Enhance Transmission to Target Cells in the Respiratory Epithelium JF - Frontiers in immunology N2 - Transmission of measles virus (MV) from dendritic to airway epithelial cells is considered as crucial to viral spread late in infection. Therefore, pathways and effectors governing this process are promising targets for intervention. To identify these, we established a 3D respiratory tract model where MV transmission by infected dendritic cells (DCs) relied on the presence of nectin-4 on H358 lung epithelial cells. Access to recipient cells is an important prerequisite for transmission, and we therefore analyzed migration of MV-exposed DC cultures within the model. Surprisingly, enhanced motility toward the epithelial layer was observed for MV-infected DCs as compared to their uninfected siblings. This occurred independently of factors released from H358 cells indicating that MV infection triggered cytoskeletal remodeling associated with DC polarization enforced velocity. Accordingly, the latter was also observed for MV-infected DCs in collagen matrices and was particularly sensitive to ROCK inhibition indicating infected DCs preferentially employed the amoeboid migration mode. This was also implicated by loss of podosomes and reduced filopodial activity both of which were retained in MV-exposed uninfected DCs. Evidently, sphingosine kinase (SphK) and sphingosine-1-phosphate (S1P) as produced in response to virus-infection in DCs contributed to enhanced velocity because this was abrogated upon inhibition of sphingosine kinase activity. These findings indicate that MV infection promotes a push-and-squeeze fast amoeboid migration mode via the SphK/S1P system characterized by loss of filopodia and podosome dissolution. Consequently, this enables rapid trafficking of virus toward epithelial cells during viral exit. KW - dendritic cell KW - cell migration KW - measles virus KW - 3D tissue model KW - sphingosine-1-phosphate Y1 - 2019 U6 - https://doi.org/10.3389/fimmu.2019.01294 SN - 1664-3224 VL - 10 PB - Frontiers Research Foundation CY - Lausanne ER - TY - JOUR A1 - Gutbier, Birgitt A1 - Schönrock, Stefanie M. A1 - Ehrler, Carolin A1 - Haberberger, Rainer A1 - Dietert, Kristina A1 - Gruber, Achim D. A1 - Kummer, Wolfgang A1 - Michalick, Laura A1 - Kuebler, Wolfgang M. A1 - Hocke, Andreas C. A1 - Szymanski, Kolja A1 - Letsiou, Eleftheria A1 - Lüth, Anja A1 - Schumacher, Fabian A1 - Kleuser, Burkhard A1 - Mitchell, Timothy J. A1 - Bertrams, Wilhelm A1 - Schmeck, Bernd A1 - Treue, Denise A1 - Klauschen, Frederick A1 - Bauer, Torsten T. A1 - Tönnies, Mario A1 - Weissmann, Norbert A1 - Hippenstiel, Stefan A1 - Suttorp, Norbert A1 - Witzenrath, Martin T1 - Sphingosine Kinase 1 Regulates Inflammation and Contributes to Acute Lung Injury in Pneumococcal Pneumonia via the Sphingosine-1-Phosphate Receptor 2 JF - Critical care medicine N2 - Objectives: Severe pneumonia may evoke acute lung injury, and sphingosine-1-phosphate is involved in the regulation of vascular permeability and immune responses. However, the role of sphingosine-1-phosphate and the sphingosine-1-phosphate producing sphingosine kinase 1 in pneumonia remains elusive. We examined the role of the sphingosine-1-phosphate system in regulating pulmonary vascular barrier function in bacterial pneumonia. Design: Controlled, in vitro, ex vivo, and in vivo laboratory study. Subjects: Female wild-type and SphK1-deficient mice, 8-10 weeks old. Human postmortem lung tissue, human blood-derived macrophages, and pulmonary microvascular endothelial cells. Interventions: Wild-type and SphK1-deficient mice were infected with Streptococcus pneumoniae. Pulmonary sphingosine-1-phosphate levels, messenger RNA expression, and permeability as well as lung morphology were analyzed. Human blood-derived macrophages and human pulmonary microvascular endothelial cells were infected with S. pneumoniae. Transcellular electrical resistance of human pulmonary microvascular endothelial cell monolayers was examined. Further, permeability of murine isolated perfused lungs was determined following exposition to sphingosine-1-phosphate and pneumolysin. Measurements and Main Results: Following S. pneumoniae infection, murine pulmonary sphingosine-1-phosphate levels and sphingosine kinase 1 and sphingosine-1-phosphate receptor 2 expression were increased. Pneumonia-induced lung hyperpermeability was reduced in SphK1(-/-) mice compared with wild-type mice. Expression of sphingosine kinase 1 in macrophages recruited to inflamed lung areas in pneumonia was observed in murine and human lungs. S. pneumoniae induced the sphingosine kinase 1/sphingosine-1-phosphate system in blood-derived macrophages and enhanced sphingosine-1-phosphate receptor 2 expression in human pulmonary microvascular endothelial cell in vitro. In isolated mouse lungs, pneumolysin-induced hyperpermeability was dose dependently and synergistically increased by sphingosine-1-phosphate. This sphingosine-1-phosphate-induced increase was reduced by inhibition of sphingosine-1-phosphate receptor 2 or its downstream effector Rho-kinase. Conclusions: Our data suggest that targeting the sphingosine kinase 1-/sphingosine-1-phosphate-/sphingosine-1-phosphate receptor 2-signaling pathway in the lung may provide a novel therapeutic perspective in pneumococcal pneumonia for prevention of acute lung injury. KW - acute lung injury KW - pneumococcal pneumonia KW - sphingosine kinase 1 KW - sphingosine-1-phosphate KW - sphingosine-1-phosphate receptor 2 Y1 - 2018 U6 - https://doi.org/10.1097/CCM.0000000000002916 SN - 0090-3493 SN - 1530-0293 VL - 46 IS - 3 SP - e258 EP - e267 PB - Lippincott Williams & Wilkins CY - Philadelphia ER - TY - JOUR A1 - Halilbasic, Emina A1 - Fuerst, Elisabeth A1 - Heiden, Denise A1 - Japtok, Lukasz A1 - Diesner, Susanne C. A1 - Trauner, Michael A1 - Kulu, Askin A1 - Jaksch, Peter A1 - Hoetzenecker, Konrad A1 - Kleuser, Burkhard A1 - Kazemi-Shirazi, Lili A1 - Untersmayr, Eva T1 - Plasma levels of the bioactive sphingolipid metabolite S1P in adult cystic fibrosis patients BT - potential target for immunonutrition? JF - Nutrients N2 - Recent research has linked sphingolipid (SL) metabolism with cystic fibrosis transmembrane conductance regulator (CFTR) activity, affecting bioactive lipid mediator sphingosine-1-phosphate (S1P). We hypothesize that loss of CFTR function in cystic fibrosis (CF) patients influenced plasma S1P levels. Total and unbound plasma S1P levels were measured in 20 lung-transplanted adult CF patients and 20 healthy controls by mass spectrometry and enzyme-linked immunosorbent assay (ELISA). S1P levels were correlated with CFTR genotype, routine laboratory parameters, lung function and pathogen colonization, and clinical symptoms. Compared to controls, CF patients showed lower unbound plasma S1P, whereas total S1P levels did not differ. A positive correlation of total and unbound S1P levels was found in healthy controls, but not in CF patients. Higher unbound S1P levels were measured in Delta F508-homozygous compared to Delta F508-heterozygous CF patients (p = 0.038), accompanied by higher levels of HDL in Delta F508-heterozygous patients. Gastrointestinal symptoms were more common in Delta F508 heterozygotes compared to Delta F508 homozygotes. This is the first clinical study linking plasma S1P levels with CFTR function and clinical presentation in adult CF patients. Given the emerging role of immunonutrition in CF, our study might pave the way for using S1P as a novel biomarker and nutritional target in CF. KW - sphingolipids KW - sphingosine-1-phosphate KW - intestine KW - high density KW - lipoproteins KW - cystic fibrosis KW - Delta F508 mutation KW - immunonutrition Y1 - 2020 U6 - https://doi.org/10.3390/nu12030765 SN - 2072-6643 VL - 12 IS - 3 PB - MDPI CY - Basel ER - TY - GEN A1 - Kleuser, Burkhard T1 - The enigma of sphingolipids in health and disease T2 - Postprints der Universität Potsdam : Mathematisch-Naturwissenschaftliche Reihe T3 - Zweitveröffentlichungen der Universität Potsdam : Mathematisch-Naturwissenschaftliche Reihe - 1040 KW - sphingosine-1-phosphate Y1 - 2020 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:kobv:517-opus4-472637 SN - 1866-8372 IS - 1040 ER - TY - GEN A1 - Plöhn, Svenja A1 - Edelmann, Bärbel A1 - Japtok, Lukasz A1 - He, Xingxuan A1 - Hose, Matthias A1 - Hansen, Wiebke A1 - Schuchman, Edward H. A1 - Eckstein, Anja A1 - Berchner-Pfannschmidt, Utta T1 - CD40 enhances sphingolipids in orbital fibroblasts BT - potential role of sphingosine-1-phosphate in inflammatory T-cell migration in Graves' orbitopathy T2 - Postprints der Universität Potsdam : Mathematisch-Naturwissenschaftliche Reihe N2 - PURPOSE. Graves' orbitopathy (GO) is an autoimmune orbital disorder associated with Graves' disease caused by thyrotropin receptor autoantibodies. Orbital fibroblasts (OFs) and CD40 play a key role in disease pathogenesis. The bioactive lipid sphingosine-1-phosphate (S1P) has been implicated in promoting adipogenesis, fibrosis, and inflammation in OFs. We investigated the role of CD40 signaling in inducing S1P activity in orbital inflammation. METHODS. OFs and T cells were derived from GO patients and healthy control (Ctl) persons. S1P abundance in orbital tissues was evaluated by immunofluorescence. OFs were stimulated with CD40 ligand and S1P levels were determined by ELISA. Further, activities of acid sphingomyelinase (ASM), acid ceramidase, and sphingosine kinase were measured by ultraperformance liquid chromatography. Sphingosine and ceramide contents were analyzed by mass spectrometry. Finally, the role for S1P in T-cell attraction was investigated by T-cell migration assays. RESULTS. GO orbital tissue showed elevated amounts of S1P as compared to control samples. Stimulation of CD40 induced S1P expression in GO-derived OFs, while Ctl-OFs remained unaffected. A significant increase of ASM and sphingosine kinase activities, as well as lipid formation, was observed in GO-derived OFs. Migration assay of T cells in the presence of SphK inhibitor revealed that S1P released by GO-OFs attracted T cells for migration. CONCLUSIONS. The results demonstrated that CD40 ligand stimulates GO fibroblast to produce S1P, which is a driving force for T-cell migration. The results support the use of S1P receptor signaling modulators in GO management. T3 - Zweitveröffentlichungen der Universität Potsdam : Mathematisch-Naturwissenschaftliche Reihe - 1099 KW - Grave’s orbitopathy KW - sphingosine-1-phosphate KW - sphingolipids KW - inflammation Y1 - 2021 U6 - http://nbn-resolving.de/urn/resolver.pl?urn:nbn:de:kobv:517-opus4-468837 SN - 1866-8372 IS - 1099 ER -